Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Angelo A. Manfredi is active.

Publication


Featured researches published by Angelo A. Manfredi.


EMBO Reports | 2004

HMGB1 is an endogenous immune adjuvant released by necrotic cells

Patrizia Rovere-Querini; Annalisa Capobianco; Paola Scaffidi; Barbara Valentinis; Federica Catalanotti; Marta Giazzon; Ingrid E Dumitriu; Susanne Müller; Matteo Iannacone; Catia Traversari; Marco Bianchi; Angelo A. Manfredi

Immune responses against pathogens require that microbial components promote the activation of antigen‐presenting cells (APCs). Autoimmune diseases and graft rejections occur in the absence of pathogens; in these conditions, endogenous molecules, the so‐called ‘innate adjuvants’, activate APCs. Necrotic cells contain and release innate adjuvants; necrotic cells also release high‐mobility group B1 protein (HMGB1), an abundant and conserved constituent of vertebrate nuclei. Here, we show that necrotic HMGB1−/− cells have a reduced ability to activate APCs, and HMGB1 blockade reduces the activation induced by necrotic wild‐type cell supernatants. In vivo, HMGB1 enhances the primary antibody responses to soluble antigens and transforms poorly immunogenic apoptotic lymphoma cells into efficient vaccines.


Nature Medicine | 2010

Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures

Mattia Maroso; Silvia Balosso; Teresa Ravizza; Jaron Liu; Eleonora Aronica; Anand M. Iyer; Carlo Rossetti; Monica Molteni; Maura Casalgrandi; Angelo A. Manfredi; Marco Bianchi; Annamaria Vezzani

Brain inflammation is a major factor in epilepsy, but the impact of specific inflammatory mediators on neuronal excitability is incompletely understood. Using models of acute and chronic seizures in C57BL/6 mice, we discovered a proconvulsant pathway involving high-mobility group box-1 (HMGB1) release from neurons and glia and its interaction with Toll-like receptor 4 (TLR4), a key receptor of innate immunity. Antagonists of HMGB1 and TLR4 retard seizure precipitation and decrease acute and chronic seizure recurrence. TLR4-defective C3H/HeJ mice are resistant to kainate-induced seizures. The proconvulsant effects of HMGB1, like those of interleukin-1β (IL-1β), are partly mediated by ifenprodil-sensitive N-methyl-d-aspartate (NMDA) receptors. Increased expression of HMGB1 and TLR4 in human epileptogenic tissue, like that observed in the mouse model of chronic seizures, suggests a role for the HMGB1-TLR4 axis in human epilepsy. Thus, HMGB1-TLR4 signaling may contribute to generating and perpetuating seizures in humans and might be targeted to attain anticonvulsant effects in epilepsies that are currently resistant to drugs.


Immunological Reviews | 2007

High-mobility group box 1 (HMGB1) protein at the crossroads between innate and adaptive immunity.

Marco Bianchi; Angelo A. Manfredi

Summary: Tissue damage occurs often in the life of mammals and is usually repaired. Dying cells are swiftly phagocytosed, but before disappearing, they alert surrounding cells to activate homeostatic programs. They release signals that recruit inflammatory cells to the site of injury, promote cell migration and cell division to replace dead cells, and activate the immune system in anticipation of microbial invasion. Many of these events involve high‐mobility group box 1 protein (HMGB1), a nuclear protein that is released passively when necrotic cells lose the integrity of their membranes. HMGB1 behaves as a trigger of inflammation, attracting inflammatory cells, and of tissue repair, recruiting stem cells and promoting their proliferation. Moreover, HMGB1 activates dendritic cells (DCs) and promotes their functional maturation and their response to lymph node chemokines. Activated leukocytes actively secrete HMGB1 in the microenvironment. Thus, HMGB1 acts in an autocrine/paracrine fashion and sustains long‐term repair and defense programs. DCs secrete HMGB1 several hours after contact with the first maturation stimulus; HMGB1 secretion is critical for their ability to reach the lymph nodes, to sustain the proliferation of antigen‐specific T cells, to prevent their activation‐dependent apoptosis, and to promote their polarization towards a T‐helper 1 phenotype. These immune responses will also be directed against self‐antigens that DCs process at the time of injury and can lead to autoimmunity.


Journal of Experimental Medicine | 2008

Induction of inflammatory and immune responses by HMGB1–nucleosome complexes: implications for the pathogenesis of SLE

Vilma Urbonaviciute; Barbara G. Fürnrohr; Silke Meister; Luis E. Munoz; Petra Heyder; Francesco De Marchis; Marco Bianchi; Carsten J. Kirschning; Hermann Wagner; Angelo A. Manfredi; Joachim R. Kalden; Georg Schett; Patrizia Rovere-Querini; Martin J. Herrmann; Reinhard E. Voll

Autoantibodies against double-stranded DNA (dsDNA) and nucleosomes represent a hallmark of systemic lupus erythematosus (SLE). However, the mechanisms involved in breaking the immunological tolerance against these poorly immunogenic nuclear components are not fully understood. Impaired phagocytosis of apoptotic cells with consecutive release of nuclear antigens may contribute to the immune pathogenesis. The architectural chromosomal protein and proinflammatory mediator high mobility group box protein 1 (HMGB1) is tightly attached to the chromatin of apoptotic cells. We demonstrate that HMGB1 remains bound to nucleosomes released from late apoptotic cells in vitro. HMGB1–nucleosome complexes were also detected in plasma from SLE patients. HMGB1-containing nucleosomes from apoptotic cells induced secretion of interleukin (IL) 1β, IL-6, IL-10, and tumor necrosis factor (TNF) α and expression of costimulatory molecules in macrophages and dendritic cells (DC), respectively. Neither HMGB1-free nucleosomes from viable cells nor nucleosomes from apoptotic cells lacking HMGB1 induced cytokine production or DC activation. HMGB1-containing nucleosomes from apoptotic cells induced anti-dsDNA and antihistone IgG responses in a Toll-like receptor (TLR) 2–dependent manner, whereas nucleosomes from living cells did not. In conclusion, HMGB1–nucleosome complexes activate antigen presenting cells and, thereby, may crucially contribute to the pathogenesis of SLE via breaking the immunological tolerance against nucleosomes/dsDNA.


Journal of Immunology | 2005

Release of High Mobility Group Box 1 by Dendritic Cells Controls T Cell Activation via the Receptor for Advanced Glycation End Products

Ingrid E. Dumitriu; Paramita Baruah; Barbara Valentinis; Reinhard E. Voll; Martin Herrmann; Peter P. Nawroth; Bernd Arnold; Marco Bianchi; Angelo A. Manfredi; Patrizia Rovere-Querini

High mobility group box 1 (HMGB1) is an abundant and conserved nuclear protein that is released by necrotic cells and acts in the extracellular environment as a primary proinflammatory signal. In this study we show that human dendritic cells, which are specialized in Ag presentation to T cells, actively release their own HMGB1 into the extracellular milieu upon activation. This secreted HMGB1 is necessary for the up-regulation of CD80, CD83, and CD86 surface markers of human dendritic cells and for IL-12 production. The HMGB1 secreted by dendritic cells is also required for the clonal expansion, survival, and functional polarization of naive T cells. Using neutralizing Abs and receptor for advanced glycation end product-deficient (RAGE−/−) cells, we demonstrate that RAGE is required for the effect of HMGB1 on dendritic cells. HMGB1/RAGE interaction results in downstream activation of MAPKs and NF-κB. The use of an ancient signal of necrosis, HMGB1, by dendritic cells to sustain their own maturation and for activation of T lymphocytes represents a profitable evolutionary mechanism.


Nature Reviews Rheumatology | 2010

The role of defective clearance of apoptotic cells in systemic autoimmunity

Luis E. Munoz; Kirsten Lauber; Martin Schiller; Angelo A. Manfredi; Martin Herrmann

The inefficient clearance of dying cells can result in the accumulation of apoptotic cell remnants. This occurrence is considered an intrinsic defect that can cause the permanent presence of cellular debris responsible for the initiation of systemic autoimmunity in diseases such as systemic lupus erythematosus (SLE). If postapoptotic debris accumulates in germinal centers, activates complement and functions as a survival signal for B cells that have become autoreactive by somatic hypermutation, autoimmunity could arise (etiology). The accumulation of postapoptotic remnants and fragments derived from secondary necrotic cells in the presence of autoantibodies against apoptotic cells or adaptor molecules obliges their pathological elimination and maintains autoinflammation. The autoimmunity that occurs in patients with SLE involves complex antigens that contain nucleic acids, which can function as virus mimetics. Complexes of autoantibodies, proteins and nucleic acids are likely to be mistaken by the immune system for opsonized viruses, resulting in the production of type I interferons, a hallmark of SLE (pathogenesis). The pathogenicity of autoantibodies is thought to strongly increase if autoantigens are accessible for immune-complex formation. The immune complex could be considered a binary pyrogen formed from less proinflammatory components. The accessibility of cognate autoantigens, in turn, is likely to be related to impaired or delayed clearance of apoptotic cells.


OncoImmunology | 2014

Consensus guidelines for the detection of immunogenic cell death

Oliver Kepp; Laura Senovilla; Ilio Vitale; Erika Vacchelli; Sandy Adjemian; Patrizia Agostinis; Lionel Apetoh; Fernando Aranda; Vincenzo Barnaba; Norma Bloy; Laura Bracci; Karine Breckpot; David Brough; Aitziber Buqué; Maria G. Castro; Mara Cirone; María I. Colombo; Isabelle Cremer; Sandra Demaria; Luciana Dini; Aristides G. Eliopoulos; Alberto Faggioni; Silvia C. Formenti; Jitka Fucikova; Lucia Gabriele; Udo S. Gaipl; Jérôme Galon; Abhishek D. Garg; François Ghiringhelli; Nathalia A. Giese

Apoptotic cells have long been considered as intrinsically tolerogenic or unable to elicit immune responses specific for dead cell-associated antigens. However, multiple stimuli can trigger a functionally peculiar type of apoptotic demise that does not go unnoticed by the adaptive arm of the immune system, which we named “immunogenic cell death” (ICD). ICD is preceded or accompanied by the emission of a series of immunostimulatory damage-associated molecular patterns (DAMPs) in a precise spatiotemporal configuration. Several anticancer agents that have been successfully employed in the clinic for decades, including various chemotherapeutics and radiotherapy, can elicit ICD. Moreover, defects in the components that underlie the capacity of the immune system to perceive cell death as immunogenic negatively influence disease outcome among cancer patients treated with ICD inducers. Thus, ICD has profound clinical and therapeutic implications. Unfortunately, the gold-standard approach to detect ICD relies on vaccination experiments involving immunocompetent murine models and syngeneic cancer cells, an approach that is incompatible with large screening campaigns. Here, we outline strategies conceived to detect surrogate markers of ICD in vitro and to screen large chemical libraries for putative ICD inducers, based on a high-content, high-throughput platform that we recently developed. Such a platform allows for the detection of multiple DAMPs, like cell surface-exposed calreticulin, extracellular ATP and high mobility group box 1 (HMGB1), and/or the processes that underlie their emission, such as endoplasmic reticulum stress, autophagy and necrotic plasma membrane permeabilization. We surmise that this technology will facilitate the development of next-generation anticancer regimens, which kill malignant cells and simultaneously convert them into a cancer-specific therapeutic vaccine.


Arthritis & Rheumatism | 2001

PTX3 in small‐vessel vasculitides: An independent indicator of disease activity produced at sites of inflammation

Fausto Fazzini; Giuseppe Peri; Andrea Doni; Giacomo Dell'Antonio; Elena Dal Cin; Enrica Bozzolo; Francesca D'Auria; Luisa Praderio; Gianfranco Ciboddo; Maria Grazia Sabbadini; Angelo A. Manfredi; Alberto Mantovani; Patrizia Rovere Querini

OBJECTIVE To verify whether the prototypical long pentraxin PTX3 represents an indicator of the activity of small-vessel vasculitis. METHODS Concentrations of PTX3, a pentraxin induced in endothelium by cytokines, were measured by enzyme-linked immunosorbent assay in the sera of 43 patients with Churg-Strauss syndrome, Wegeners granulomatosis, and microscopic polyangiitis. PTX3 was also measured in the sera of 28 patients with systemic lupus erythematosus (SLE), 22 with rheumatoid arthritis, and 16 with CREST syndrome (calcinosis, Raynauds phenomenon, esophageal dysmotility, sclerodactyly, telangiectasias). Serum concentrations of C-reactive protein (CRP) were measured by immunoturbidimetry. The cells involved in PTX3 production in vivo were identified in skin biopsy samples. RESULTS Patients with active vasculitis had significantly higher concentrations of PTX3 than did those with quiescent disease (P < 0.001). PTX3 levels in the latter group were similar to those in healthy controls. PTX3 levels were higher in patients with untreated vasculitis and lower in patients who underwent immunosuppressive treatments (P < 0.005). In contrast, patients with active SLE had negligible levels of the pentraxin. PTX3 levels did not correlate with CRP levels in vasculitis patients. Endothelial cells produced PTX3 in active skin lesions. CONCLUSION PTX3 represents a novel acute-phase reactant produced at sites of active vasculitis.


Journal of Leukocyte Biology | 2007

The secretion of HMGB1 is required for the migration of maturing dendritic cells

Ingrid E. Dumitriu; Marco Bianchi; Monica Bacci; Angelo A. Manfredi; Patrizia Rovere-Querini

Chemokines regulate the migration and the maturation of dendritic cells (DC) licensed by microbial constituents. We have recently found that the function of DC, including their ability to activate naïve, allogeneic CD4+ T cells, requires the autocrine/pracrine release of the nuclear protein high mobility group box 1 (HMGB1). We show here that human myeloid DC, which rapidly secrete upon maturation induction their own HMGB1, remodel their actin‐based cytoskeleton, up‐regulate the CCR7 and the CXCR4 chemokine receptors, and acquire the ability to migrate in response to chemokine receptor ligands. The events are apparently causally related: DC challenged with LPS in the presence of HMGB1‐specific antibodies fail to up‐regulate the expression of the CCR7 and CXCR4 receptors and to rearrange actin‐rich structures. Moreover, DC matured in the presence of anti‐HMGB1 antibodies fail to migrate in response to the CCR7 ligand CCL19 and to the CXCR4 ligand CXCL12. The blockade of receptor for advanced glycation end products (RAGE), the best‐characterized membrane receptor for HMGB1, impinges as well on the up‐regulation of chemokine receptors and on responsiveness to CCL19 and CXCL12. Our data suggest that the autocrine/paracrine release of HMGB1 and the integrity of the HMGB1/RAGE pathway are required for the migratory function of DC.


European Journal of Immunology | 2005

Requirement of HMGB1 and RAGE for the maturation of human plasmacytoid dendritic cells

Ingrid E. Dumitriu; Paramita Baruah; Marco Bianchi; Angelo A. Manfredi; Patrizia Rovere-Querini

Dendritic cells (DC) are key components of innate and adaptive immune responses. Plasmacytoid DC (PDC) are a specialized DC subset that produce high amounts of type I interferons in response to microbes. High mobility group box 1 protein (HMGB1) is an abundant nuclear protein, which acts as a potent pro‐inflammatory factor when released extracellularly. We show that HMGB1 leaves the nucleus of maturing PDC following TLR9 activation, and that PDC express on the plasma membrane the best‐characterized receptor for HMGB1, RAGE. Maturation and type I IFN secretion of PDC is hindered when the HMGB1/RAGE pathway is disrupted. These results reveal HMGB1 and RAGE as the first known autocrine loop modulating the maturation of PDC, and suggest that antagonists of HMGB1/RAGE might have therapeutic potential for the treatment of systemic human diseases.

Collaboration


Dive into the Angelo A. Manfredi's collaboration.

Top Co-Authors

Avatar

Patrizia Rovere-Querini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Maria Grazia Sabbadini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Norma Maugeri

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Giuseppe A. Ramirez

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Marco Bianchi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Enrico Tombetti

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Elena Baldissera

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Annalisa Capobianco

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Clara Sciorati

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Antonella Monno

Vita-Salute San Raffaele University

View shared research outputs
Researchain Logo
Decentralizing Knowledge